EP2457578B1 - Molécule chimère comprenant angiopoietine-1 et une domaine superhélice pour le traitement de la dysfonction érectile pénienne - Google Patents

Molécule chimère comprenant angiopoietine-1 et une domaine superhélice pour le traitement de la dysfonction érectile pénienne Download PDF

Info

Publication number
EP2457578B1
EP2457578B1 EP11008503.2A EP11008503A EP2457578B1 EP 2457578 B1 EP2457578 B1 EP 2457578B1 EP 11008503 A EP11008503 A EP 11008503A EP 2457578 B1 EP2457578 B1 EP 2457578B1
Authority
EP
European Patent Office
Prior art keywords
ang1
comp
coiled
domain
receptor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP11008503.2A
Other languages
German (de)
English (en)
Other versions
EP2457578A1 (fr
Inventor
Gou Young Koh
Ghung-Hyun Cho
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aprogen Inc
Original Assignee
Aprogen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aprogen Inc filed Critical Aprogen Inc
Publication of EP2457578A1 publication Critical patent/EP2457578A1/fr
Application granted granted Critical
Publication of EP2457578B1 publication Critical patent/EP2457578B1/fr
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/515Angiogenesic factors; Angiogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/73Fusion polypeptide containing domain for protein-protein interaction containing coiled-coiled motif (leucine zippers)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor

Definitions

  • the present invention provides for novel applications of chimeric coiled-coil molecule such as COMP-Ang1 for recovering artherosclerotic erectile dysfunction.
  • Angiopoietin family proteins include four different angiopoietins, namely, angiopoietin-1 (Ang1), angiopoietin-2 (Ang2), angiopoietin-3 (Ang3) and angiopoietin-4 (Ang4) ( Yancopoulos et al., 2000, Nature 407:242-245 ; Koh et al., 2002, Exp Mol Med. 34:1-11 ) ( Fig . 1 ).
  • Ang1 angiopoietin-1
  • Ang2 angiopoietin-2
  • Ang3 angiopoietin-3
  • Ang4 angiopoietin-4
  • Ang1 and Ang4 act as agonist to vascular endothelial cell specific receptor tyrosine kinase, Tie2, while Ang2 and Ang3 have been known to act as antagonist to Tie2 ( Yancopoulos et al., 2000, Nature 407:242-248 ; Koh et al., 2002, Exp Mol Med. 34:1-11 ) ( Fig . 1 ).
  • Ang2 and Ang3 also can act as agonist in context dependent manner ( Kim et al., Oncogene. 200014:4549-4552 ; Lee et al., FASEB J. 18:1200-1208 ) ( Fig. 1 ).
  • Ang1 Biochemical organization of Ang1 is complicated.
  • Schematic diagram of the protein structure of Ang1 indicates that amino acids 1-19 are the secretory signal sequences (S), amino acids 20-155 are the superclustering domain (SCD), amino acids 156-255 are the coiled-coil oligomeric domain (CCOD), amino acids 256-283 are the linker (L), and amino acids 284-498 are the fibrinogen-like domain (FLD) ( Fig. 2 ).
  • cysteines (C) at amino acids 41, 54, 265, 286, 315, 435, 437, 439, and 452 ( Fig, 2 ).
  • COMP-Ang1 For in vivo use of Ang1, a soluble, stable, and potent Ang1 variant, COMP-Ang1 was developed ( Cho et al., PNAS 101:5547-5552, 2004 ; Cho et al., PNAS 101:5553-5558, 2004 ; US patent 7,081,443 and WO2003/106501 ). COMP-Ang1 is more potent than native Ang1 in phosphorylating the Tie2 receptor in lung endothelial cells in vitro and in vivo ( Cho et al., PNAS 101 5547-5552, 2004 ; Cho et al., PNAS 101:5553-5558, 2004 ; US patent 7,081,443 and WO2003/106501 ).
  • WO2003/048185 discloses making various fusion proteins with angioprotein, but does not discuss uses of the protein for treating penile erectile dysfunction.
  • WO2003/034990 discloses angiogenic factors and in particular angiopoietin 1 to induce vasodilation and reduce hypertension.
  • the reference mentions the condition of penile erectile dysfunction and hypothesizes that Ang1 may be useful to treat this type of condition
  • this reference provides no data or any substantive evidence that any compound disclosed in the this reference can be reasonably used to treat penile erectile dysfunction.
  • COMP-Ang1 produces enhanced healthy, non-leaky and stable angiogenesis, lymphangiogenesis and blood flow during recovery of penile erectile dysfunction in adult animal.
  • COMP-Ang1 produces (1) specific increase number of blood vessels, (2) specific enlargement of blood vessels, (3) specific increase of lymphatic vessels, without overt pathologic changes (within physiological ranges) in the adult animals.
  • Vascular enlargement is a characteristic feature of angiopoietin-1 (Ang1)-induced changes in adult blood vessels.
  • Ang1 angiopoietin-1
  • COMP-Ang1 a soluble, stable and potent Ang1 variant
  • COMP-Ang1 recombinant protein induced transient vascular enlargement that spontaneously reversed within a month.
  • the vascular enlargement resulted from endothelial proliferation.
  • the COMP-Ang1-induced vascular remodeling is mainly mediated through Tie2 activation.
  • Sustained overexpression of Tie2 could participate in the maintenance of vascular changes.
  • the present invention is directed to treating a variety of diseases for which angiogenesis could be the treatment modality.
  • the coiled coil domain may be from cartilage matrix protein or cartilage oligomeric matrix protein, and further the receptor binding domain may bind to Tie1 or Tie2.
  • the ligand may be a cytokine, hormone or growth factor such as an angiopoietin-related protein, including angiopoietin-related protein-1 (ARP1), angiopoietin-related protein-2 (ARP2), hepatic fibrinogen/angiopoietin-related protein (HFARP).
  • ARP1 angiopoietin-related protein-1
  • ARP2 angiopoietin-related protein-2
  • HFARP hepatic fibrinogen/angiopoietin-related protein
  • the ligand may be angiopoietin-1, angiopoietin-2, angiopoietin-3 or angiopoietin-4, and in particular, angiopoietin-1.
  • the chimeric coiled coil molecule is COMP-Ang1.
  • the ischemic disease may be, but is not limited to, a disease of the heart, limb, brain, penis, stomach or kidney.
  • the administration of the chimeric coiled coil molecule may be carried out by administering the protein form or by gene construct form, as popularly termed gene therapy for long lasting delivery of the chimeric coiled coil molecule.
  • the gene delivery vehicle may be any method including liposomes, viruses, such as retrovirus or adnovirus or virus like particles, such as adeno-associated viruses.
  • the invention is directed to treating penile erectile dysfunction comprising administering to a person in need thereof an effective amount of a coiled coil chimeric molecule comprising a coiled-coil domain linked to a receptor binding domain of a ligand.
  • the erectile dysfunction may be artherosclerotic or vascular diseased type. The method of treatment using chimeric coiled coil molecule and protein and gene delivery are discussed above.
  • Figure 1 shows the interaction between angiopoietins and Tie2.
  • Angiopoietins bind to endothelial cell specific receptor kinase Tie2 and Tie1 and are known to regulate vasculogenesis, angiogenesis and lymphangiogeneis, and to maintain endothelial cell integrity.
  • Ang1 and Ang3 act as agonists
  • Ang2 and Ang4 act as not only agonists but also act as antagonists in a context dependent manner.
  • FIG 2 shows a schematic diagram outlining the protein structure of native Ang1.
  • Amino acids 1-19 are the secretory signal sequence (S)
  • amino acids 20-158 are the superclustering domain (SCD)
  • amino acids 158-255 are the coiled-coil oligomeric domain (CCOD)
  • amino acids 226-283 are the linker (L)
  • amino acids 284-498 are the fibrinogen-like domain (FLD).
  • cysteines (C) at amino acids 41, 54, 265, 315, 435, 437,439, and 452.
  • Figures 3A-3F show the effect of VEGF and COMP-Ang1 in punched-hole injury in the ear of various ages. Indicated ages of male FVB/n mice were treated with 1x10 9 pfu Ade-LacZ (Control, A and D ), 1x10 8 pfu Ade-mVEGF (mouse VEGF, B and E ) or 1x10 9 pfu Ade-COMP-Ang1 ( C and F ) virus, and a closed punched-hole injury was made in the ear. At indicated times or days later, ears were photographed.
  • Figure 4 shows survival rate of mice treated with VEGF and COMP-Ang1. Eight week-old male FVB/n mice were treated with indicated amount of Ade-mVEGF or Ade-COMP-Ang1 virus, and survival rate was measured.
  • Figures 5A-5D show the effect of COMP-Ang1 on vascular remodeling in ear skin.
  • Eight week-old male FVB/N mice were treated with daily injection of 200 ⁇ g of BSA ( A and C ) or 200 ⁇ g of COMP-Ang1 recombinant protein ( B and D ) for 14 days.
  • Sections of ear skin were stained with H&E ( A and B ) and blood vessels of whole- mounted ear skin were visualized with PECAM-1 (CD31) immunostaining (red) ( C and D ).
  • the mice treated with COMP-Ang1 have prominently enlarged blood vessels (red arrow-heads in H&E staining) in the ear skin.
  • FIGS 6A-6B show the effect of COMP-Ang1 on vascular remodeling in abdominal skin.
  • Vascular specific expression of eGFP using Tie2 promoter transgenic mice were treated with daily injection of 200 ⁇ g of BSA (A) or 200 ⁇ g of COMP-Ang1 recombinant protein ( B ) for 14 days.
  • Blood vessels of abdominal skin sections were visualized with PECAM-1 (CD31) immunostaining (red).
  • the mice treated with COMP-Ang1 have prominently enlarged blood vessels.
  • Figures 7A-7B show the effect of adenoviral COMP-Ang1 on skin color.
  • Eight week-old male BALB/c- nu nude mice were treated with 1x10 9 pfu Ade-LacZ (Control, A ) or Ade-COMP-Ang1 ( B ). Two weeks later, the skin color of the face, hands, and upper trunk were photographed. The mice treated with Ade-CONT-Ang1 show overt skin redness. Results from 3 experiments were similar.
  • Figures 8A-8B show the effect of adenoviral COMP-Ang1 on skin color.
  • Eight week-old male BALB/c- nu nude mice were treated with 1x10 9 pfu Ade-LacZ (Control, A ) or Ade-COMP-Ang1 ( B ).
  • Five weeks later, blood vessels of ear skin were photographed.
  • the mice treated with Ade-COMP-Ang1 show striking increase in number, size and branching patterns of blood vessels in the ear compared to control-treated mice.
  • FIGS 9A-9F show the effect of adenoviral COMP-Ang1 on ischemic hindlimb.
  • Ischemic hindlimb mouse model was generated in eight week-old male BALB/c- nu nude mice by partial ligation of femoral artery. Then, 100 ⁇ g of BSA (A, B, C) or 100 ⁇ g of COMP-Ang1 ( D, E, F ) was injected directly into ischemic muscles at day 0 ( A and D ), 3 (B and E) and 5 ( C and F ). Indicated times, blood flow in the ischemic legs were measued by laser micro-Doppler method. Compared to BSA treatment, there is almost complete recovery of blood flow in the ischemic hindlimb in COMP-Ang1 treated mice.
  • Figures 10A-10F show the effect of COMP-Ang1 on corpora cavernosal sinus of penis.
  • Eight week-old male FVB/N mice were treated with 1x10 9 pfu Ade-LacZ ( A and D ) or Ade-COMP-Ang1 ( B, C, E and F ). Indicated times later, sections of penis were immunostained with anti-von-Willebrand factor antibody (pinkish violet). A, B and C, magnification x40; D, E and F, magnification x200.
  • the penis of mouse treated with Ade-COMP-Ang1 shows enlarged corpora cavernosal sinus with induction of von-Willebrand factor in a time dependent manner.
  • Figures 11A-11B show the effect of COMP-Ang1 on corpora cavernosal sinus of penis. Eight weeks-old male FVB/N mice were treated with treated with 1x10 9 pfu Ade-LacZ ( A ) or 1x10 9 pfu Ade-COMP-Ang1 ( B ). Three weeks later, epoxy sections of penis were stained with toluidine blue. The penis of mouse treated with Ade-COMP-Ang1 shows enlarged corpora cavernosal sinus.
  • Figures 12A-12F show the effect of COMP-Ang1 on corpora cavernosal sinus of penis.
  • Eight week-old male FVB/N mice were treated with 1x10 9 pfu Ade-LacZ ( A, B and C ) or 1x10 9 pfu Ade-COMP-Ang1 ( D, E and F ).
  • Three weeks later, sections of penis were immunostained with anti-PECAM-1 antibody (red) or anti-von-Willebrand factor antibody (red).
  • the penis of mouse treated with Ade-COMP-Ang1 shows enlarged corpora cavernosal sinus with induction of von-Willibrand factor in a time dependent manner.
  • Figures 13A-13C show the protocol of generation of hypercholesterolemic erectile dysfunctional rat model and treatment of COMP-Ang1.
  • A Eight-week old male Sprague-Dawley rat were fed with 4% cholesterol plus 1% cholic acid diet for 3 months. Then, adenoviral COMP-Ang1 (5x10 7 pfu) or adenoviral LacZ (5x10 7 pfu) was injected directly into corpora cavernosum of hypercholesterolemic erectile dysfunctional rat ( B and C ).
  • Figures 14A-14E show the effect of COMP-Ang1 on erectile function in hypercholesterolemic erectile dysfunctional rat.
  • ICP intra-corporal pressure
  • SAP systemic arterial pressure
  • Figures 15A-15D show the effect of COMP-Ang1 on penis of hypercholesterolemic erectile dysfunctional rat.
  • sections of penis were immunostained with anti-von-Willebrand factor antibody (pinkish violet).
  • the penis of mouse treated with COMP-Ang1 shows enlarged corpora cavernosal sinus with induction of von-Willebrand factor in a dose-dependent manner.
  • Figures 16A-16B show the effect of COMP-Ang1 on penis of hypercholesterolemic erectile dysfunctional rat.
  • Fifty ⁇ g of BSA ( A ) or COMP-Ang1 ( B ) recombinant protein was injected directly into corpora cavernosum of hypercholesterolemic erectile dysfunctional rat as every alternative day for 4 times.
  • sections of penis were immunostained with anti-endothelial nitric oxide synthase (eNOS) antibody (red).
  • eNOS anti-endothelial nitric oxide synthase
  • the penis treated with COMP-Ang1 shows induction of eNOS ( A ).
  • a method of using coiled-coil domains for "multimerizing" ligands which enhances the biological activity of such ligands that, absent such multimerization, would have lower levels of biological activity.
  • This method may be used to multimerize receptor binding domains from any ligand that has improved affinity and/or increased activity (i.e. signaling ability) when they were multimerized as compared to the non-multimerized form of the ligand.
  • the present invention also provides for methods of using coiled-coil domains for "multimerizing" soluble receptors, which functions to make otherwise inactive soluble receptors biologically active, or which enhances the biological and binding activity of receptors that, absent such multimerization, would have lower levels of biological and binding activity.
  • This method may be used to multimerize ligand binding domains using any receptor, which has improved affinity and/or increased activity (i.e. binding) when they were multimerized as compared to the native form of the soluble receptor.
  • polypeptide As used herein, “about” or “substantially” generally provides a leeway from being limited to an exact number. For example, as used in the context of the length of a polypeptide sequence, “about” or “substantially” indicates that the polypeptide is not to be limited to the recited number of amino acids. A few amino acids add to or subtracted from the N-terminus or C-terminus may be included so long as the functional activity such as its binding activity is present.
  • administration "in combination with” one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order.
  • agonist refers to a ligand that binds to a receptor, which activates the receptor and stimulates physiologic activity.
  • Ang1 is considered to be an agonist of Tie2 receptor.
  • amino acid and “amino acids” refer to all naturally occurring L- ⁇ -amino acids. This definition is meant to include norleucine, ornithine, and homocysteine.
  • amino acid sequence variant refers to molecules with some differences in their amino acid sequences as compared to a reference (e.g. native sequence) polypeptide.
  • the amino acid alterations may be substitutions, insertions, deletions or any desired combinations of such changes in a native amino acid sequence.
  • Substitutional variants are those that have at least one amino acid residue in a native sequence removed and a different amino acid inserted in its place at the same position.
  • the substitutions may be single, where only one amino acid in the molecule has been substituted, or they may be multiple, where two or more amino acids have been substituted in the same molecule.
  • Substitutes for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs.
  • the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan and methionine.
  • the polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine and glutamine.
  • the positively charged (basic) amino acids include arginine, lysine and histidine.
  • the negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • proteins or fragments or derivatives thereof which exhibit the same or similar biological activity and derivatives which are differentially modified during or after translation, e.g., by glycosylation, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, and so on.
  • Insertional variants are those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a native amino acid sequence. Immediately adjacent to an amino acid means connected to either the ⁇ -carboxy or ⁇ -amino functional group of the amino acid.
  • Deletional variants are those with one or more amino acids in the native amino acid sequence removed. Ordinarily, deletional variants will have one or two amino acids deleted in a particular region of the molecule.
  • antagonist refers to a ligand that tends to nullify the action of another ligand, as a ligand that binds to a cell receptor without eliciting a biological response.
  • biologically active refers to the ability of a molecule to specifically bind to and signal through a native receptor, e.g., a native Tie2 receptor, or to block the ability of a native Tie receptor (e.g., Tie2) to participate in signal transduction.
  • the (native and variant) ligands of the present invention include agonists and antagonists of a native receptor, e.g. Tie2 receptor.
  • Preferred biological activities of the ligands of the present invention include the ability to induce or inhibit vascularization. The ability to induce vascularization will be useful for the treatment of biological conditions and disease, where vascularization is desirable. On the other hand, the ability to inhibit or block vascularization may, for example, be useful in preventing or attenuating cell proliferation and tumor growth.
  • Preferred biological activities of the ligands of the present invention include the ability to inhibit vascular permeability.
  • the ability to inhibit vascular permeability will be useful for treatment of medical conditions and diseases such as diabetic retinopathy, edema, and ascites.
  • Preferred biological activities of the ligands of the present invention include the ability to maintain endothelial cell integrity (including preventing apoptosis). The ability to maintain endothelial cell integrity will be useful for treatment of medical conditions and diseases such as mannitol treatment, irradiation, and sepsis.
  • the biological activity of the chimeric receptor includes its ability to inhibit or competitively inhibit the ligand's activity by binding to its ligand.
  • cell proliferation may be inhibited if the ligand is an agonist for cell proliferation.
  • administration of chimeric receptor may act as an enhancer of cell proliferation if the ligand is an antagonist for cell proliferation.
  • chimeric ligand and chimeric receptor be labeled with a detectable label, such as radioisotope, fluorescent tag, enzymatic tag, or a chemiluminescent tag to determine ligand-receptor binding interaction.
  • a detectable label such as radioisotope, fluorescent tag, enzymatic tag, or a chemiluminescent tag to determine ligand-receptor binding interaction.
  • assay systems employing the chimeric molecule is also contemplated.
  • carriers include pharmaceutically acceptable carriers, excipients, or stabilizers which are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed.
  • pharmaceutically acceptable carrier is an aqueous pH buffered solution.
  • Examples of pharmaceutically acceptable carriers include without limitation buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; saltforming counterions such as sodium; and/or nonionic surfactants such as TWEEN ® , polyethylene glycol (PEG), and PLURONICS ® .
  • buffers such as phosphate, citrate, and other organic acids
  • antioxidants including ascorbic acid
  • low molecular weight (less than about 10 residues) polypeptide such
  • chimeric coiled coil molecule and “coiled coil chimeric molecule” are used interchangeably.
  • chimeric ligand refers to the combination of coiled coil domain and a receptor binding domain or a ligand binding domain.
  • the resultant chimeric polypeptide is capable of forming biologically active multimers, which are soluble.
  • the coiled coil domain may be derived from any source, including any animal or mammalian protein, and in particular any human protein, and further includes those that are synthetically made.
  • the coiled coil domain and the ligand or receptor constructs may be from the same or different source.
  • the chimeric construct comprises the coiled coil domain and a receptor binding domain of a ligand or a ligand binding domain of a receptor, and further may include other components that may be included so long their inclusion does not interfere with the formation of a biologically active multimer that has improved solubility, ease of recombinant production of the chimeric polypeptide and substantially similar or greater potency as the native ligand or native soluble receptor.
  • FLAG sequence may be included for ease of purification, provided its inclusion does not interfere with the function of the chimeric molecule.
  • the FLAG sequence also may be removed if a humanized construct is desired.
  • an effective amount is an amount sufficient to effect beneficial or desired clinical or biochemical results.
  • An effective amount can be administered one or more times.
  • an effective amount of an inhibitor compound is an amount that is sufficient to palliate, ameliorate, stabilize, reverse, slow or delay the progression of the disease state.
  • fragments or “functional derivatives” refers to biologically active amino acid sequence variants and fragments of the native ligands or receptors of the present invention, as well as covalent modifications, including derivatives obtained by reaction with organic derivatizing agents, post-translational modifications, derivatives with nonproteinaceous polymers, and immunoadhesins.
  • host cell includes an individual cell or cell culture which can be or has been a recipient of a vector of this invention.
  • Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation and/or change.
  • ligand refers to any molecule or agent, or compound that specifically binds covalently or transiently to a molecule such as a polypeptide.
  • ligand may include antibody.
  • ligand may refer to a molecule sought to be bound by another molecule with high affinity, such as in a ligand trap.
  • ligand binding domain refers to the portion of the receptor that binds to the ligand and includes the minimal portion of the receptor that is necessary to bind its ligand.
  • linked refers to direct or indirect connection between the multimerizing domain and the ligand or receptor. Both a direct fusion between these two domains or indirect fusion as by the domains being separated by a linker or an intervening domain or element are contemplated, so long as the activity of the chimeric fusion is present.
  • mammal for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, cats, cattle, horses, sheep, pigs, and so on.
  • the mammal is human.
  • multimer or “multimeric” refers to the joining of the multimerizing agent such as the coiled coil domain to each other to form a dimer, trimer, tetramer, pentamer, hexamer, heptamer, octamer, nanomer, decamer and so on, which may be in a parallel or anti-parallel form, through intramolecular or intermolecular bonds.
  • pharmaceutically acceptable carrier and/or diluent includes any and all solvents, dispersion media, coatings antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, use thereof in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active material for the treatment of disease in living subjects having a diseased condition in which bodily health is impaired.
  • the principal active ingredient is compounded for convenient and effective administration in effective amounts with a suitable pharmaceutically acceptable carrier in dosage unit form.
  • a unit dosage form can, for example, contain the principal active compound in amounts ranging from 0.5 ⁇ g to about 2000 mg. Expressed in proportions, the active compound is generally present in from about 0.5 ⁇ g/ml of carrier. In the case of compositions containing supplementary active ingredients, the dosages are determined by reference to the usual dose and manner of administration of the said ingredients.
  • receptor binding domain refers to the portion of the ligand that binds to the receptor and includes the minimal portion of the ligand that is necessary to bind its receptor.
  • the present invention is based on the discovery that a multimerizing agent, such as a coiled coil domain, which was previously perceived as a source of hindrance for isolating recombinant proteins containing them, has been found to provide advantageous features of easy recombinant protein expression and purification, greater solubility and greater or substantially equal potency compared with the native protein containing the coiled coil domain.
  • sample or “biological sample” is referred to in its broadest sense, and includes any biological sample obtained from an individual, body fluid, cell line, tissue culture, or other source which may contain a chimeric Ang1 binding factor, depending on the type of assay that is to be performed.
  • biological samples include body fluids, such as semen, lymph, sera, plasma, urine, synovial fluid, spinal fluid and so on. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art.
  • the term “specifically binds” refers to a non-random binding reaction between two molecules, for example between an antibody molecule immunoreacting with an antigen, or a non-antibody ligand reacting with another polypeptide, such as chimeric Ang1 specifically binding with Tie2.
  • subject is a vertebrate, preferably a mammal, more preferably a human.
  • treatment is an approach for obtaining beneficial or desired clinical results.
  • beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Treatment refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • “Palliating" a disease means that the extent and/or undesirable clinical manifestations of a disease state are lessened and/or the time course of the progression is slowed or lengthened, as compared to a situation without treatment.
  • a polynucleotide vector of this invention may be in any of several forms, including, but not limited to, RNA, DNA, RNA encapsulated in a retroviral coat, DNA encapsulated in an adenovirus coat, DNA packaged in another viral or viral-like form (such as herpes simplex, and adeno-associated virus (AAV)), DNA encapsulated in liposomes, DNA complexed with polylysine, complexed with synthetic polycationic molecules, complexed with compounds such as polyethylene glycol (PEG) to immunologically "mask” the molecule and/or increase half-life, or conjugated to a non-viral protein.
  • RNA Ribonucleic acid
  • the polynucleotide is DNA.
  • DNA includes not only bases A, T, C, and G, but also includes any of their analogs or modified forms of these bases, such as methylated nucleotides, intemucleotide modifications such as uncharged linkages and thioates, use of sugar analogs, and modified and/or alternative backbone structures, such as polyamides.
  • the present invention includes a multimer forming domain.
  • coiled coil domain is exemplified.
  • the coiled coil domain may be any amino acid sequence that forms a coiled coil structure. While the exemplified coiled coil domains herein are those cloned from a variety of proteins, it is understood that various mutations and derivatization are encompassed by the invention, so long as the resultant coiled coil domain is recognized by a person of skill in the art as a coiled coil structure and the coiled coil domain containing chimera is capable of forming a multimer, is easily soluble, and is able to provide similar or greater potency with respect to the native ligand or receptor.
  • the multimerizing domain and the binding domain may be from the same protein, or they may be from different proteins.
  • Ang1 coiled coil domain may be linked to its own fibrinogen-like domain in a more efficient manner.
  • a cartilage oligomeric matrix protein could be linked to the Ang1 fibrinogen-like domain.
  • Ang1 portion referred to is the fibrinogen domain of Ang1
  • MAT-Ang1 is also sometimes referred to as CMP-Ang1, CMP-Ang1/FD, or CMP/CC-Ang1/FD in the figures.
  • Ang1 COMP-Ang1 has many potential advantages over the native protein.
  • the generation of native Ang1 is difficult, and the activities of the purified proteins vary, possibly due to its tendency to form multimers.
  • Our structural analysis of native Ang1 by rotary shadowing TEM indicates that native Ang1 exists as variably-sized multimers.
  • MAT-Ang1 and COMP-Ang1 yielded additional oligomers. Nevertheless, MAT-Ang1 and COMP-Ang1 are easier to purify and more soluble than native Ang1.
  • COMP-Ang1 is approximately 3-5 times more potent than native Ang1.
  • COMP-Ang1 produced the most potent effect on Tie2 and Akt phosphorylation.
  • COMP-Ang1 could be the most biologically active among the variants because of its rapid association and dissociation rate.
  • COUP-Ang1 may induce Tie2 clustering in endothelial caveolae more efficiently than the other recombinant Ang1 proteins.
  • Tie2 molecules are localized in endothelial caveolae.
  • an engineered Ang1 protein with an oligomeric structure better suited to clustering Tie2 in caveolae may better facilitate Tie2 multimerization and phosphorylation.
  • Ang1 failed to stimulate an angiogenic response when administered alone.
  • Ang1 augmented postnatal angiogenesis.
  • native Ang1 failed to stimulate an angiogenic response.
  • COMP-Ang1 alone stimulates angiogenesis.
  • COMP-Ang1 produced an increase in the luminal diameter of the basal limbus. This role is consistent with increased frequency of enlarged vessel diameters in Ang1-overexpressing transgenic mice ( Suri, C. et al., 1998, Science 282: 468-471 ; Thurston, G. et al., 1999, Science 286:2511-2514 ).
  • COMP-Ang1 can produce more effective blood flow by increasing the diameter of arterial lumens.
  • Ang1 can counteract VEGF-induced side effects such as edema and inflammation, while having an additive effect on angiogenesis ( Yamak, H. J. et al., 2000, Circulation 101:2317-2324 ; Thurston, G. el al., 1999, Science 286:2511-2514 ; Kim, I. et al., 2001, Circ. Res. 89:477-479 ).
  • COMP-Ang1 protein delivery may be useful for accurate and safe therapeutic angiogenesis.
  • ⁇ -helical coiled coil is probably the most widespread subunit oligomerization motif found in proteins. Accordingly, coiled coils fulfill a variety of different functions. In several families of transcriptional activators, for example, short leucine zippers play an important role in positioning the DNA-binding regions on the DNA ( Ellenberger et al., 1992, Cell 71:1223-1237 ). Coiled coils are also used to form oligomers of intermediate filament proteins. Coiled-coil proteins furthermore appear to play an important role in both vesicle and viral membrane fusion ( Skehel and Wiley, 1998, Cell 95:871-874 ).
  • hydrophobic sequences embedded in the membranes to be fused, are located at the same end of the rod-shaped complex composed of a bundle of long ⁇ -helices. This molecular arrangement is believed to cause close membrane apposition as the complexes are assembled for membrane fusion.
  • the coiled coil is often used to control oligomerization. It is found in many types of proteins, including transcription factors such as, but not limited to GCN4, viral fusion peptides, SNARE complexes and certain tRNA synthetases, among others. Very long coiled coils are found in proteins such as tropomyosin, intermediate filaments and spindle-pole-body components.
  • Coiled coils involve a number of ⁇ -helices that are supercoiled around each other in a highly organized manner that associate in a parallel or an antiparallel orientation. Although dimers and trimers are the most common.
  • the helices may be from the same or from different proteins.
  • the coiled-coil is formed by component helices coming together to bury their hydrophobic seams. As the hydrophobic seams twist around each helix, so the helices also twist to coil around each other, burying the hydrophobic seams and forming a supercoil. It is the characteristic interdigitation of side chains between neighbouring helices, known as knobs-into-holes packing, that defines the structure as a coiled coil.
  • the helices do not have to run in the same direction for this type of interaction to occur, although parallel conformation is more common. Antiparallel conformation is very rare in trimers and unknown in pentamers, but more common in intramolecular dimers, where the two helices are often connected by a short loop.
  • the heterotrimeric coiled-coil protein laminin plays an important role in the formation of basement membranes.
  • Other examples are the thrombospondins and cartilage oligomeric matrix protein (COMP) in which three (thrombospondins I and 2) or five (thrombospondins 3, 4 and COMP) chains are connected.
  • the molecules have a flower bouquet-like appearance, and the reason for their oligomeric structure is probably the multivalent interaction of the C-terminal domains with cellular receptors.
  • the yeast transcriptional activator GCN4 is 1 of over 30 identified eukaryotic proteins containing the basic region leucine zipper (bZIP) DNA-binding motif ( Ellenberger et al., 1992, Cell 71:1223-1237 ).
  • the bZIP dimer is a pair of continuous alpha helices that form a parallel coiled-coil over their carboxy-terminal 34 residues and gradually diverge toward their amino termini to pass through the major groove of the DNA binding site.
  • the coiled-coil dimerization interface is oriented almost perpendicular to the DNA axis, giving the complex the appearance of the letter T.
  • bZIP contains a 4-3 heptad repeat of hydrophobic and nonpolar residues that pack together in a parallel alpha-helical coiled-coil ( Ellenberger et al., 1992, Cell 71:1223-1237 ).
  • the stability of the dimer results from the side-by-side packing of leucines and nonpolar residues in positions a and d of the heptad repeat, as well as a limited number of intra- and interhelical salt bridges, shown in a crystal structure of the GCN4 leucine zipper peptide ( Ellenberger et al., 1992, Cell 71:1223-1237 ).
  • CMP Cartilage Matrix Protein
  • CMP (matrilin-1) was isolated from bovine tracheal cartilage as a homotrimer of subunits of M r 52,000 ( Paulsson and Heineg ⁇ rd, 1981, Biochem J. 197:367-375 ), where each subunit consists of a vWFAI module, a single EGF domain, a vWFA2 module and a coiled coil domain spanning five heptads ( Kiss et al., 1989, J. Biol. Chem. 264:8126-8134 ; Hauser and Paulsson, 1994, J. Biol. Chem. 269:25747-25753 ).
  • Electron microscopy of purified CMP showed a bouquet-like trimer structure in which each subunit forms an ellipsoid emerging from a common point corresponding to the coiled coil ( Hauser and Paulsson, 1994, J. Biol. Chem. 269:25747-25753 ).
  • the coiled coil domain in matrilin-I has been extensively studied.
  • the trimeric structure is retained after complete reduction of interchain disulfide bonds under non-denaturing conditions ( Hauser and Paulsson, 1994, J. Biol. Chem. 269:25747-25753 ).
  • COMP non-collagenous glycoprotein
  • the protein is a 524 kDa homopentamer of five subunits which consists of an N-terminal heptad repeat region (cc) followed by four epidermal growth factor (EGF)-like domains (EF), seven calcium-binding domains (T3) and a C-terminal globular domain (TC).
  • EGF epidermal growth factor
  • T3 calcium-binding domains
  • TC C-terminal globular domain
  • COMP belongs to the family of thrombospondins.
  • Tie2 receptor antagonist fusion polypeptides are used to diagnose or treat patients in which the desired result is inhibition of a hematopoietic pathway, such as for the treatment of myeloproliferative or other proliferative disorders of blood forming organs such as thrombocythemias, polycythemias and leukemias.
  • treatment may comprise use of a therapeutically effective amount of the fusion polypeptides as described herein.
  • compositions for use according to the invention include the fusion polypeptides described above in a pharmacologically acceptable liquid, solid or semi-solid carrier, linked to a carrier or targeting molecule (e.g., antibody, hormone, growth factor, etc.) and/or incorporated into liposomes, microcapsules, and controlled release preparation prior to administration in vivo.
  • a carrier or targeting molecule e.g., antibody, hormone, growth factor, etc.
  • the pharmaceutical composition may comprise a fusion polypeptide in an aqueous solution, such as sterile water, saline, phosphate buffer or dextrose solution.
  • the active agents may be comprised in a solid (e.g. wax) or semi-solid (e.g. gelatinous) formulation that may be implanted into a patient in need of such treatment
  • the administration route may be any mode of administration known in the art, including but not limited to intravenously, intrathecally, subcutaneously, intrauterinely, by injection into involved tissue, intraarterially, intranasally, orally, or via an implanted device.
  • Administration may result in the distribution of the active agent of the invention throughout the body or in a localized area.
  • intravenous or intrathecal administration of agent may be desirable.
  • an implant containing active agent may be placed in or near the lesioned area.
  • Suitable implants include, but are not limited to, gelfoam, wax, spray, or microparticle-based implants.
  • compositions comprising the fusion polypeptides described herein, in a pharmacologically acceptable vehicle.
  • the compositions may be administered systemically or locally. Any appropriate mode of administration known in the art may be used, incuding, but not limited to, intravenous, intrathecal, intraarterial, intranasal, oral, subcutaneous, intraperitoneal, or by local injection or surgical implant. Sustained release formulations are also provided for.
  • nucleic acids comprising sequences encoding the chimeric Ang1 polypeptide are administered to prevent vascular leakage, and for therapeutic vasculogenesis, by way of gene therapy.
  • Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acids produce their encoded protein that mediates a therapeutic effect.
  • nucleic acid sequences may encode a chimeric-Ang1 or Tie2 polypeptide, in which the nucleic acid sequences are part of expression vectors that express the polypeptides in a suitable host.
  • such nucleic acid sequences have promoters operably linked to the polypeptide coding region, said promoter being inducible or constitutive, and, optionally, tissue-specific.
  • nucleic acid molecules are used in which the polypeptide coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the antibody encoding nucleic acids ( Koller and Smithies, Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989 ); Zijlstra et al., Nature 342:435-438 (1989 ).
  • Delivery of the nucleic acids into a patient may be either direct, in which case the patient is directly exposed to the nucleic acid or nucleic acid- carrying vectors, or indirect, in which case, cells are first transformed with the nucleic acids in vitro, then transplanted into the patient. These two approaches are known, respectively, as in vivo or ex vivo gene therapy.
  • the nucleic acid sequences are directly administered in vivo , where it is expressed to produce the encoded product.
  • This can be accomplished by any of numerous methods known in the art, e.g., by constructing them as part of an appropriate nucleic acid expression vector and administering it so that they become intracellular, e.g., by infection using defective or attenuated retrovirals or other viral vectors, or by direct injection of naked DNA, or coating with lipids or cell-surface receptors or transfecting agents, encapsulation in liposomes, microparticles, or microcapsules, or by administering them in linkage to a peptide which is known to enter the nucleus, by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see, e.g., Wu and Wu, J.
  • nucleic acid-ligand complexes can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor.
  • the nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination ( Koller and Smithies, Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989 ); Zijlstra et al., Nature 342:435-438 (1989 )).
  • viral vectors that contain nucleic acid sequences encoding the polypeptide are used.
  • the nucleic acid sequences encoding the polypeptide to be used in gene therapy are cloned into one or more vectors, which facilitates delivery of the gene into a patient.
  • Retroviral vectors, adenoviral vectors and adeno-associated viruses are examples of viral vectors that may be used. Retroviral vectors contain the components necessary for the correct packaging of the viral genome and integration into the host cell DNA.
  • Adenoviruses are especially attractive vehicles for delivering genes to respiratory epithelia because they naturally infect respiratory epithelia where they cause a mild disease.
  • Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle.
  • Adenoviruses have the advantage of being capable of infecting non-dividing cells.
  • adeno-associated virus AAV has also been proposed for use in gene therapy.
  • Another approach to gene therapy involves transferring a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection.
  • the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transferred gene. Those cells are then delivered to a patient.
  • the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell.
  • introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, spheroplast fusion and so on.
  • Numerous techniques are known in the art for the introduction of foreign genes into cells and may be used in accordance with the present invention, provided that the necessary developmental and physiological functions of the recipient cells are not disrupted.
  • the technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
  • Cells into which a nucleic acid can be introduced for purposes of gene therapy encompass any desired, available cell type, and include but are not limited to epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T-lymphocytes, B-lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord blood, peripheral blood, fetal liver, and so on.
  • the cell used for gene therapy is autologous to the patient.
  • nucleic acid sequences encoding the polypeptide are introduced into the cells such that they are expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect.
  • stem or progenitor cells are used. Any stem and/or progenitor cells which can be isolated and maintained in vitro can potentially be used in accordance with this embodiment of the present invention.
  • the nucleic acid to be introduced for purposes of gene therapy comprises an inducible promoter operably linked to the coding region, such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
  • the present invention relates to treatment for various diseases that are characterized by being treatable through therapeutic angiogenesis such as but not limited to vascular leakage or lack of blood vessel formation.
  • inventive therapeutic compound may be administered to human patients who are either suffering from, or prone to suffer from the disease by providing compounds that activate Tie2.
  • the formulation of therapeutic compounds is generally known in the art and reference can conveniently be made to Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa., USA .
  • Dosage regime may be adjusted to provide the optimum therapeutic response.
  • several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • the active compound may be administered in a convenient manner such as by the oral, intravenous (where water soluble), intramuscular, subcutaneous, intra nasal, intradermal or suppository routes or implanting (eg using slow release molecules by the intraperitoneal route or by using cells e.g.
  • the peptide may be required to be coated in a material to protect it from the action of enzymes, acids and other natural conditions which may inactivate said ingredients.
  • the low lipophilicity of the peptides will allow them to be destroyed in the gastrointestinal tract by enzymes capable of cleaving peptide bonds and in the stomach by acid hydrolysis.
  • they will be coated by, or administered with, a material to prevent its inactivation.
  • peptides may be administered in an adjuvant, co-administered with enzyme inhibitors or in liposomes.
  • Adjuvants contemplated herein include resorcinols, non-ionic surfactants such as polyoxyethylene oleyl ether and n-hexadecyl polyethylene ether.
  • Enzyme inhibitors include pancreatic trypsin inhibitor, diisopropylfluorophosphate (DEP) and trasylol.
  • Liposomes include water-in-oil-in-water CGF emulsions as well as conventional liposomes.
  • the active compounds may also be administered parenterally or intraperitoneally.
  • Dispersions can also be prepared in glycerol liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of superfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, chlorobutanol, phenol, sorbic acid, theomersal and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the composition of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterile active ingredient into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and the freeze-drying technique which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the active compound may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet.
  • the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 1% by weight of active compound.
  • compositions and preparations may, of course, be varied and may conveniently be between about 5 to about 80% of the weight of the unit.
  • the amount of active compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • Preferred compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains between about 0.1 ⁇ g and 2000 mg of active compound.
  • the tablets, pills, capsules and the like may also contain the following: A binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin may be added or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring.
  • a binder such as gum tragacanth, acacia, corn starch or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin may be added or a flavoring agent such as peppermint, oil of winter
  • tablets, pills, or capsules may be coated with shellac, sugar or both.
  • a syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor.
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and formulations.
  • a compound of the invention e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis, construction of a nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • a protein including an antibody or a peptide of the invention
  • care must be taken to use materials to which the protein does not absorb.
  • the compound or composition can be delivered in a vesicle, in particular a liposome.
  • the compound or composition can be delivered in a controlled release system.
  • a pump may be used.
  • polymeric materials can be used.
  • a controlled release system can be placed in proximity of the therapeutic target, thus requiring only a a fraction of the systemic dose.
  • COMP-Ang1 may be useful for treating patients with ischemic organ disease including heart, limbs, brain, and stomach. Therefore, we generated ischemic hindlimb mouse model by partial ligation of femoral artery and followed up blood flow in the ischemic area by laser micro-Doppler method. Then, adenoviral COMP-Ang1 (5x10 7 pfu) or adenoviral LacZ (5x10 7 pfu) was injected directly into ischemic muscles. Compared to adenoviral LacZ treatment, there was almost complete recovery of blood flow in the ischemic hindlimb of the adenoviral COMP-Ang1 treated mice ( Fig. 9 ). Therefore, COMP-Ang1 is useful for treating patients with ischemic diseases.
  • COMP-Ang1 may be useful for treating patients with atherosclerotic erectile dysfunction. Therefore, we generated hypercholesterolemic erectile dysfunctional rat model by feeding 4% cholesterol plus 1% cholic acid diet for 3 months ( Fig. 13 ). Then, adenoviral COMP-Ang1 (5x10 7 pfu) or adenoviral LacZ (5x10 7 pfu) was injected directly into corpora cavernosum of hypercholesterolemic erectile dysfunctional rat ( Fig. 13 ).
  • COMP-Ang1 a soluble, stable and potent Ang1 variant
  • COMP-Ang1 Cho et al., 2004, Proc Natl Acad Sci USA. 101:5547-52 .
  • COMP-Ang1 is more potent than native Ang1 in phosphorylating the Tie2 receptor and signaling via Akt in primary cultured endothelial cells ( Cho et al., 2004, Proc Natl Acad Sci USA. 101:5547-52 ).
  • Tie1 an endothelial-specific receptor tyrosine kinase, shares a high degree of homology with Tie2. Although Tie1 was isolated over a decade ago ( Partanen et al., 1992, Mol Cell Biol. 12:1698-707 ), no ligand had been found to activate it. Recently, Saharinen et al ., demonstrated that COMP-Ang1 stimulated Tie1 phosphorylation in cultured endothelial cells ( Saharinen et al., 2005, J Cell Biol. 169:239-243 ).
  • COMP-Ang1-induced Tie1 activation was amplified via Tie2 and was more efficient than native Ang1- and Ang4-induced Tie1 activation.
  • COMP-Ang1 and Ang1 are now known to be activating ligands for both Tie1 and Tie2.
  • COMP-Ang1-induced vascular remodeling in adult tracheal vessels is mainly mediated through activation of Tie2, not by Tie1.
  • Ang1-induced vessel enlargement is a unique characteristic among many growth factors.
  • Our immunohistological examination of phosphohistone H3 revealed that COMP-Ang1-induced vascular enlargements were evidently the result of endothelial proliferation, which is consistent with a recent report ( Baffert et al., 2004, Circ Res. 94:984-992 ).
  • arteriolar and venular enlargements are achieved mainly by circumferential endothelial proliferation, which is a unique phenomenon and is different from multi-directional endothelial cell proliferation during vasculogenesis and angiogenesis.
  • our results revealed that different organs show different sensitivities to long-term and sustained COMP-Ang1.
  • COMP-Ang1 could provide a great therapeutic benefit to patients with delayed skin wound healing and ischemic heart diseases through its ability to promote vascular remodeling.
  • the mice treated with long-lasting and sustained COMP-Ang1 did not show any significant changes in body weight, systemic blood pressure, or heart rate. More detailed analysis will be necessary to clarify how it is possible that the mice with enlarged blood vessels caused by long-term and sustained COMP-Ang1 have a normal blood pressure and heart rate.
  • Ang1 is known to be a strong growth factor for pericyte recruitment to nascent endothelial cells during development ( Suri et al., 1996, Cell. 87:1171-1180 ; Suri et al., 1998, Science. 282:468- 471 ; Thurston et al., 1999, Science. 286:2511-2514 ).
  • This Ang1-induced pericyte recruitment is related to the Ang1-induced anti-leakage effect on VEGF and pro-inflammatory stimuli ( Thurston et al., 1999, Science. 286:2511-2514 ).
  • our results show a lower number and poorer covering of pericytes in COMP-Ang1-induced enlarged postcapillary venules.
  • Ang1 is able to restore a hierarchical architecture of growing blood vessels and rescues retinal edema and hemorrhage even in the absence of pericyte recruitment ( Uemura et al., 2002. J Clin Invest 110:1619-1628 ).
  • COMP-Ang1 may be able to assemble endothelial cells in a frame of hierarchical architecture without pericyte recruitment in the COMP-Ang1-induced enlarged blood vessels.
  • Recombinant Chinese hamster ovary (rCHO) cells expressing COMP-Ang1 (CA1-2; production rate, -30 mg/L) were established as previously described ( Hwang et al., 2005, Protein Express Purif. 39:175-183 ).
  • Recombinant adenovirus expressing COMP-Ang1 or LacZ was constructed using the pAdEasy TM vector system (Qbiogene).
  • EXAMPLE 1.2 Animals, treatment and measurement of blood pressure and heart rate
  • FVB/N mice and Tie2-GFP transgenic mice were purchased from Jackson Laboratory and bred in our pathogen-free animal facility. Male mice 8-10 weeks old were used for this study. Animal care and experimental procedures were performed under approval from the Animal Care Committees of KAIST. For protein treatment, 200 ⁇ g of COMP-Ang1 recombinant protein or BSA dissolved in 50 ⁇ L of sterile 0.9% NaCl was injected daily through the tail vein for 2 weeks.
  • Ade-COMP-Angl Ade-LacZ
  • Ade-sTie2-Fc soluble Tie2 receptor adenovirus construct

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Diabetes (AREA)
  • Vascular Medicine (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Endocrinology (AREA)
  • Obesity (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Dermatology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Emergency Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Reproductive Health (AREA)
  • Otolaryngology (AREA)

Claims (6)

  1. Utilisation d'une quantité efficace d'une molécule chimère en superhélice comprenant un domaine de superhélice de la protéine matricielle oligomérique du cartilage (COMP) lié à un domaine de liaison au récepteur Tie2 qui est un domaine de type fibrinogène de l'angiopoïétine-1 pour la fabrication d'un médicament destiné au traitement de la dysfonction érectile pénienne.
  2. Utilisation selon la revendication 1, dans laquelle la dysfonction érectile est athérosclérotique ou de type maladie vasculaire.
  3. Utilisation selon la revendication 1, dans laquelle la molécule chimère en superhélice doit être administrée sous la forme d'un produit de recombinaison génique au sein d'un véhicule de délivrance.
  4. Utilisation selon la revendication 1, dans laquelle la molécule chimère en superhélice est un multimère soluble biologiquement actif.
  5. Utilisation selon la revendication 4, dans laquelle le multimère est un dimère, un trimère, un tétramère, un pentamère, un hexamère, un heptamère, un octamère, un nanomère ou un décamère.
  6. Utilisation selon la revendication 1, dans laquelle la molécule chimère en superhélice doit être administrée directement dans le corps caverneux.
EP11008503.2A 2004-09-28 2005-09-28 Molécule chimère comprenant angiopoietine-1 et une domaine superhélice pour le traitement de la dysfonction érectile pénienne Not-in-force EP2457578B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US61413604P 2004-09-28 2004-09-28
EP05824800A EP1819355A4 (fr) 2004-09-28 2005-09-28 Methodes d'utilisation d'une molecule superhelice chimere

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
EP05824800A Division EP1819355A4 (fr) 2004-09-28 2005-09-28 Methodes d'utilisation d'une molecule superhelice chimere
EP05824800.6 Division 2005-09-28

Publications (2)

Publication Number Publication Date
EP2457578A1 EP2457578A1 (fr) 2012-05-30
EP2457578B1 true EP2457578B1 (fr) 2015-08-19

Family

ID=36119260

Family Applications (2)

Application Number Title Priority Date Filing Date
EP05824800A Withdrawn EP1819355A4 (fr) 2004-09-28 2005-09-28 Methodes d'utilisation d'une molecule superhelice chimere
EP11008503.2A Not-in-force EP2457578B1 (fr) 2004-09-28 2005-09-28 Molécule chimère comprenant angiopoietine-1 et une domaine superhélice pour le traitement de la dysfonction érectile pénienne

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP05824800A Withdrawn EP1819355A4 (fr) 2004-09-28 2005-09-28 Methodes d'utilisation d'une molecule superhelice chimere

Country Status (7)

Country Link
US (4) US7691365B2 (fr)
EP (2) EP1819355A4 (fr)
JP (3) JP4943336B2 (fr)
KR (1) KR100980693B1 (fr)
CA (1) CA2581769C (fr)
DK (1) DK2457578T3 (fr)
WO (1) WO2006035319A2 (fr)

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006035319A2 (fr) * 2004-09-28 2006-04-06 Genexel-Sein, Inc. Methodes d'utilisation d'une molecule superhelice chimere
KR101482483B1 (ko) 2006-04-07 2015-01-15 에르피오 세러퓨틱스 인코포레이티드 인간 단백질 타이로신 포스파타아제 베타(hptp베타)에 결합하는 항체 및 그의 용도
US7622593B2 (en) 2006-06-27 2009-11-24 The Procter & Gamble Company Human protein tyrosine phosphatase inhibitors and methods of use
AU2010271105C1 (en) 2009-01-12 2014-08-21 Aerpio Therapeutics, Inc. Compounds, compositions, and methods for preventing metastasis of cancer cells
EP2397547A4 (fr) 2009-02-10 2012-09-05 Univ Ryukyus Transporteur de médicament et adjuvant et vaccin l'utilisant chacun
CA2797247C (fr) 2010-04-28 2019-10-22 Sunnybrook Health Sciences Centre Procedes et utilisations d'agents de liaison a tie2 et/ou d'agents d'activation de tie2
CN104039351A (zh) 2011-10-13 2014-09-10 阿尔皮奥治疗学股份有限公司 用于治疗血管渗漏综合征和癌症的方法
KR101407730B1 (ko) * 2012-11-23 2014-06-12 인하대학교 산학협력단 안지오포이에틴-4 단백질을 유효성분으로 포함하는 발기부전의 예방 또는 치료용 조성물
US20150050277A1 (en) 2013-03-15 2015-02-19 Aerpio Therapeutics Inc. Compositions and methods for treating ocular diseases
EP2983695B1 (fr) 2013-04-11 2019-05-29 Sunnybrook Research Institute Méthodes, utilisations et compositions d'agonistes de tie2
JP2016537416A (ja) 2013-11-01 2016-12-01 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. 脳マラリアを治療するためのアンギオポエチンに基づいた介入
JP6483148B2 (ja) 2014-03-14 2019-03-13 エアーピオ セラピューティクス インコーポレイテッド HPTP−β阻害剤
US20170304367A1 (en) * 2014-10-29 2017-10-26 University Of Utah Research Foundation Methods of treating hypoxia-associated optical conditions with cartilage oligo matrix protein-angiopoietin 1 (comp-ang1)
US20160215289A1 (en) * 2015-01-22 2016-07-28 The Regents Of The University Of California Methods of modulating lymphangiogenesis, e.g., to treat corneal transplant rejection, in a subject
PL3328886T3 (pl) * 2015-07-29 2021-02-08 Allergan, Inc. Przeciwciała ang-2 o wyłącznie ciężkim łańcuchu
KR20180054677A (ko) 2015-09-23 2018-05-24 에르피오 세러퓨틱스 인코포레이티드 Tie-2의 활성화제로 안내압을 치료하는 방법
GB202108761D0 (en) 2021-06-18 2021-08-04 Ikarovec Ltd Ocular disorders
KR20230046999A (ko) 2021-09-29 2023-04-06 주식회사 엔바이오스 코일드 코일 융합 단백질

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4810643A (en) 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US5332671A (en) 1989-05-12 1994-07-26 Genetech, Inc. Production of vascular endothelial cell growth factor and DNA encoding same
JP2657113B2 (ja) 1989-10-16 1997-09-24 アムジエン・インコーポレーテツド 幹細胞因子
EP0550296A3 (en) 1991-11-28 1993-12-29 Terumo Corp Vascular endothelial cells growth factor
US5922572A (en) 1994-01-25 1999-07-13 Human Genome Sciences, Inc. Polynucleotides encoding haemopoietic maturation factor
EP0821728B1 (fr) 1995-04-06 2004-08-11 Regeneron Pharmaceuticals, Inc. Ligands tie-2, procedes d'obtention de ces ligands et leurs utilisations
WO2003034990A2 (fr) * 2001-10-25 2003-05-01 Regeneron Pharmaceuticals, Inc. Angiopoietines et methodes d'utilisation
WO2003048185A2 (fr) * 2001-11-30 2003-06-12 Genvec, Inc. Facteurs associes a l'angiopoietine
US7081443B2 (en) * 2002-05-21 2006-07-25 Korea Advanced Institutes Of Science And Technology (Kaist) Chimeric comp-ang1 molecule
GB2409456B (en) * 2003-10-30 2006-01-04 Proimmune Ltd Oligomeric receptor ligand pair member complexes
WO2006035319A2 (fr) * 2004-09-28 2006-04-06 Genexel-Sein, Inc. Methodes d'utilisation d'une molecule superhelice chimere

Also Published As

Publication number Publication date
JP5746006B2 (ja) 2015-07-08
DK2457578T3 (en) 2015-12-07
JP4943336B2 (ja) 2012-05-30
KR20070074578A (ko) 2007-07-12
CA2581769A1 (fr) 2006-04-06
CA2581769C (fr) 2014-03-11
US20120251503A1 (en) 2012-10-04
US8206698B2 (en) 2012-06-26
JP2015091850A (ja) 2015-05-14
WO2006035319A2 (fr) 2006-04-06
US20140073566A1 (en) 2014-03-13
US20100256042A1 (en) 2010-10-07
JP2008514585A (ja) 2008-05-08
EP1819355A2 (fr) 2007-08-22
JP6018648B2 (ja) 2016-11-02
US7691365B2 (en) 2010-04-06
KR100980693B1 (ko) 2010-09-07
US8414877B2 (en) 2013-04-09
EP1819355A4 (fr) 2009-08-12
JP2012082207A (ja) 2012-04-26
US20070298996A1 (en) 2007-12-27
EP2457578A1 (fr) 2012-05-30
WO2006035319A3 (fr) 2006-06-08

Similar Documents

Publication Publication Date Title
EP2457578B1 (fr) Molécule chimère comprenant angiopoietine-1 et une domaine superhélice pour le traitement de la dysfonction érectile pénienne
EP1549679B1 (fr) Proteine de fusion comprenant un domaine se liant au recepteur de l'angiopoietine et un domaine de multimerisation
AU2008296733B2 (en) VEGFR-1/NRP-1 targeting peptides
KR102100420B1 (ko) 표적특이적으로 물질을 전달하는 엑소좀의 제조방법 및 상기 방법으로 제조된 엑소좀
IL141141A (en) A pooled polypeptide that includes a collagen binding domain and a blood vessel formation regulating domain
EP2704736A2 (fr) Procédés et compositions de ciblage de cellules adipeuses chez des mammifères
JP2019518786A (ja) インターロイキンの組み合せ及びその使用
ES2248998T3 (es) Utilizacion de las osteoprotegerina para la prevencion y el tratamiento de enfermedades cardiovasculares.
JPH11510686A (ja) TGF−βスーパーファミリーの一本鎖類縁体(モーフォン)
WO2018219301A1 (fr) VARIANT DE LIGAND INDUISANT L'APOPTOSE LIÉ AU FACTEUR DE NÉCROSE DE TUMEURS CIBLÉ PAR PDGFRβ, SON PROCÉDÉ DE PRÉPARATION ET SON UTILISATION
US11357863B2 (en) Peptide conjugates
KR20110013365A (ko) 세포투과성 엔도스타틴 재조합 단백질, 이를 코딩하는 폴리뉴클레오티드 및 이를 유효성분으로 함유하는 항암 조성물
CN109503719B (zh) 一种plg和crd功能片段重组融合蛋白及其制备方法和抗癌药物
KR20200107842A (ko) 트레일 트라이머와 암표적 펩타이드를 멀티디스플레이하는 페리틴 나노케이지 및 이의 항암제로서의 용도

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AC Divisional application: reference to earlier application

Ref document number: 1819355

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

17P Request for examination filed

Effective date: 20121129

17Q First examination report despatched

Effective date: 20130107

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20150227

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AC Divisional application: reference to earlier application

Ref document number: 1819355

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 743314

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150915

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602005047314

Country of ref document: DE

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

REG Reference to a national code

Ref country code: CH

Ref legal event code: PCOW

Free format text: NEW ADDRESS: 545 DUNCHON-DAERO (B1 HALLA SIGMA VALLEY, SANGDAEWON-DONG) JUNGWON-GU, SEONGNAM-SI,, GYEONGGI-DO, 132 15 (KR)

Ref country code: CH

Ref legal event code: NV

Representative=s name: KELLER AND PARTNER PATENTANWAELTE AG, CH

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602005047314

Country of ref document: DE

Representative=s name: FIENER, JOSEF, DE

Ref country code: DE

Ref legal event code: R081

Ref document number: 602005047314

Country of ref document: DE

Owner name: APROGEN, INC., SEONGNAM-SI, KR

Free format text: FORMER OWNER: APROGEN, INC., DAEJON, YUSEONG-GU, KR

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20151130

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 743314

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150819

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151120

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151221

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151219

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602005047314

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

26N No opposition filed

Effective date: 20160520

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150928

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 12

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20050928

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 13

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150819

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150928

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20180911

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20180913

Year of fee payment: 14

Ref country code: SE

Payment date: 20180911

Year of fee payment: 14

Ref country code: DK

Payment date: 20180911

Year of fee payment: 14

Ref country code: GB

Payment date: 20180926

Year of fee payment: 14

Ref country code: NL

Payment date: 20180919

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20181130

Year of fee payment: 14

REG Reference to a national code

Ref country code: DE

Ref legal event code: R231

Ref document number: 602005047314

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF THE APPLICANT RENOUNCES

Effective date: 20190824

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20190930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190929

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20191001

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20191001

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190930

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190930

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20190928

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190928

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190930

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190930